Skip to main content
Erschienen in: Journal of Inflammation 1/2012

Open Access 01.12.2012 | Research

Induction of intestinal pro-inflammatory immune responses by lipoteichoic acid

verfasst von: Mojgan Zadeh, Mohammad W Khan, Yong Jun Goh, Kurt Selle, Jennifer L Owen, Todd Klaenhammer, Mansour Mohamadzadeh

Erschienen in: Journal of Inflammation | Ausgabe 1/2012

Abstract

Background

The cellular and molecular mechanisms of inflammatory bowel disease are not fully understood; however, data indicate that uncontrolled chronic inflammation induced by bacterial gene products, including lipoteichoic acid (LTA), may trigger colonic inflammation resulting in disease pathogenesis. LTA is a constituent glycolipid of Gram-positive bacteria that shares many inflammatory properties with lipopolysaccharide and plays a critical role in the pathogenesis of severe inflammatory responses via Toll-like receptor 2. Accordingly, we elucidate the role of LTA in immune stimulation and induced colitis in vivo.

Methods

To better understand the molecular mechanisms utilized by the intestinal microbiota and their gene products to induce or subvert inflammation, specifically the effect(s) of altered surface layer protein expression on the LTA-mediated pro-inflammatory response, the Lactobacillus acidophilus s urface l ayer p rotein (Slp) genes encoding SlpB and SlpX were deleted resulting in a SlpB- and SlpX- mutant that continued to express SlpA (assigned as NCK2031).

Results

Our data show profound activation of dendritic cells by NCK2031, wild-type L. acidophilus (NCK56), and purified Staphylococcus aureus-LTA. In contrary to the LTA-deficient strain NCK2025, the LTA-expressing strains NCK2031 and NCK56, as well as S. aureus-LTA, induce pro-inflammatory innate and T cell immune responses in vivo. Additionally, neither NCK2031 nor S. aureus-LTA supplemented in drinking water protected mice from DSS-colitis, but instead, induced significant intestinal inflammation resulting in severe colitis and tissue destruction.

Conclusions

These findings suggest that directed alteration of two of the L. acidophilus NCFM-Slps did not ameliorate LTA-induced pro-inflammatory signals and subsequent colitis.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-9255-9-7) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MZ and MWK carried out all animal work, immunocytochemistry (i.e., high throughput and confocal imaging) and flow cytometry experiments. Additionally, MZ and MWK gathered the raw data and using statistical analyses and graphic programs compiled the data into meaningful information. MZ, MWK, JLO also contributed to writing the Methods section of the manuscript. MZ and MWK collected all of the graphics and formed the figures of the Manuscript. YJG, TK designed experiments and contributed to writing the Methods section. KS performed Slp proteome analysis on NCK2031 strain. MM designed experiments, discussed and wrote the manuscript. All authors have read and approved this manuscript.
Abkürzungen
DCs
Dendritic cells
LTA
Lipoteichoic acid
IL
Interleukin
TNFα
Tumor Necrosis Factor-alpha
Slps
Surface layer proteins
DAI
Disease Activity Index.

Background

The intestinal immune system must co-exist with resident commensal microorganisms while maintaining the ability to defend against potential microbial challenge. This immune tolerance is a highly regulated process comprised of a myriad of biological checkpoints necessary to maintain homeostasis between the host and the gut microbiota [1]. In instances of inflammatory bowel disease (IBD), this tolerance between immune cells and intestinal bacteria is disrupted; however, causes of this tolerance breakdown have not yet been determined [2, 3]. Although the etiology of IBD is still unknown, exaggerated inflammation induced by activated innate immune cells via their interaction with the microbiota and their gene products, as well as infiltrating CD4+ IFNγ+ T cells, likely play key roles in uncontrolled inflammation and tissue destruction [46]. Foxp3+ regulatory T cells (Tregs) also critically control intestinal inflammation [7] and significantly prevent colitis [8], suggesting a pivotal role for Tregs in intestinal immune homeostasis [9].
A fundamental challenge in preventing an imbalanced immune response is the understanding of how the host immune system distinguishes a pathogen from normal intestinal flora. One of the commensal microorganisms of the gut is L. acidophilus, which expresses unique s urface l ayer p roteins (Slps), including A, B, X, and abundant lipoteichoic acid (LTA). LTA is a zwitterionic glycolipid found in the cell wall of several Gram-positive bacterial strains, including L. acidophilus, which facilitates the adhesion, colonization, and invasion of cells by the bacteria [10, 11]. The best studied form of LTA is composed of a polyglycerophosphate chain that is tethered to the membrane via a glycolipid anchor [12]. Studies indicate that LTA shares many of the inflammatory properties of lipopolysaccharide (LPS) via interactions with Toll-like receptors (TLRs) [1316] which evoke diverse responses in innate cells through distinct signaling cascades [17].
Previously, we have demonstrated that deletion of the gene responsible for LTA biosynthesis in L. acidophilus NCFM diminishes this bacterium's capacity to stimulate the immune system; thereby suppressing pathogenic CD4+T cells in induced colitis [18, 19]. To further investigate the role of LTA in inflammation, we engineered the NCK2031 strain in order to evaluate the effects, if any, of altered s urface l ayer p rotein (Slp) expression on LTA-induced pro-inflammatory signals and colitis.

Methods

Materials Six to 8-week-old C57BL/6 were purchased from Jackson Laboratories (Bar Harbor, ME). Mice were maintained in microisolator cages under specific pathogen-free, Helicobacter- free conditions. Experiments were performed in an accredited establishment according to NIH guidelines in the Guide for Care and Use of Laboratory Animals (NIH-72-23), and animal protocols were approved by the local ethics committee. Dextran Sulfate Sodium (DSS) was obtained from MP Biochemicals (Solon, OH). Monoclonal antibodies for CD4, CD25, CD3, CD11c, CD11b, CD40, CD44, CD80, CD83, CD86, CD103, IL-10, IL-12, IFNγ, TNFα, HLA-ABC (R&D systems, Minneapolis, MN) and (BD, Franklin Lakes, NJ), CD1a (Dako, Carpentaria, CA), mouse and human GM-CSF and IL-4 were purchased from Invitrogen (Carlsbad, CA).
Generation of NCK2031 To generate an L. acidophilus NCFM isogenic mutant defective in all three slp genes (slpA, slpB, and slpX), the slpB (LBA0175) and slpX (LBA0512) genes were sequentially deleted in an NCFMΔupp background host (NCK1909) using the upp-based counterselective gene replacement system [20]. Subsequently, attempts to insertionally inactivate the slpA gene (LBA0169) were made within the ΔslpBX double mutant (NCK2030) using a pORI-based gene knockout system [21]. The resulting ΔslpBX strain was deficient in SlpB and SlpX, but due to genetic instability of the insertion vector, NCK2031 continued to express SlpA (data not shown). Subsequently, wild-type L. acidophilus (NCK56), NCK2031, or LTA-deficient NCK2025 were propagated in de Man, Rogosa, and Sharpe broth (MRS, Difco) at 37°C for 15 hrs. The concentration of each L. acidophilus strain was adjusted to 1 × 109 CFU/ml based on OD600 readings that had previously been correlated with CFU numbers [22]. Each mouse was orally treated with 5 × 108 CFU bacterial strain; therefore 500 μl of 1 × 109 CFU/ml suspension was centrifuged, pelleted and then resuspended in 100 μl of PBS. Each mouse was then treated with 100 μl PBS (control group) or 5 × 108 CFU (of either L. acidophilus strain) orally in100 μl sterile PBS. Fecal pellets were collected from before, during, and for up to 8 days after L. acidophilus oral treatments. Each fecal pellet was resuspended in PBS (1:10 dilution, w/v). The suspension was then serially diluted and plated onto MRS agar containing Em (2 μg/mL). The homogenized material was serially diluted and plated onto MRS agar containing Em (2 μg/mL). The daily average CFU of the L. acidophilus strains in mouse feces were determined. For in vitro stimulation, bone marrow or human monocyte derived DCs were stimulated at a 1:1, 1:10, 1:100 ratio with live NCK2031, NCK56, NCK2025, or S. aureus-LTA. Subsequently the ratio of 1:1 was chosen, as this did not overwhelmingly activate DCs and therefore did not result in cell anergy or apoptosis. For the oral gavage of mice, each mouse received 5 × 108 CFU of NCK2031, NCK56, or NCK2025 in 100 μL of PBS.
Cell culture Immature human [22, 23] or murine DCs [24] were treated with live L. acidophilus strains cells at 1:1 or with S. aureus-LTA (50 μg/mL) for 24 hrs. L. acidophilus- or S. aureus-LTA treated and untreated DCs (5 × 105) were then stained and analyzed by BD FACSCaliber or a multicolor FACSCanto. Cell supernatants were also collected and analyzed using cytometric bead array kits (BD Biosciences). At least 1 × 104 gated events per condition were acquired. Analysis software (BD CellQuest) allowed for calculation of cytokine values in supernatants at pg/mL.
Immunofluorescence The colons of mice (n = 5/group) treated with live NCK2031, NCK56, NCK2025, S. aureus-LTA, or PBS alone were fillet-opened, rolled, and snap frozen at -80°C in Tissue-Tek O.C.T. (Sakura Finetek USA, Inc., Torrance, CA). Sections (5 μm) were cut, fixed in ice-cold methanol (-20°C) for 15 min and blocked with 1% BSA. Sections were then incubated overnight at 4°C with purified hamster anti-mouse CD11c (BD Biosciences) and rat anti-mouse IL-10 (BioLegend), and antibodies for IL-12, IFNγ, TNFα, CD8, and Foxp3, followed by washing twice with PBS and incubation with anti-hamster AlexaFluor 594 and anti-rat AlexaFluor 488 (Invitrogen) for 1 hr. Sections were then washed twice with PBS and incubated for 10 minutes with 4,6-diamidino-2-phenylindole dihydrochloride (DAPI, Invitrogen), washed with PBS two times and mounted with anti-fade mounting medium, as described previously [25]. Images were acquired using TissueGnostics Tissue/Cell High Throughput Imaging and Analysis System and analyzed using ImageJ software.
Flow cytometry Groups of mice (5 mice/group) were inoculated orally with NCK2031, NCK56, NCK2025, or S. aureus-LTA (5 × 108 CFU/100 μL of sterile PBS/mouse) or PBS alone. Mice were then sacrificed after 1, 3, or 7 days; single cells were isolated from the colons [18], MLNs, and spleens and stained with CD11c, CD11b, CD103, MHC II, CD40, CD80, CD86, F4/80, CD4, CD3, CD80, or CD44 [26]. Stained cells were then fixed, permeabilized, and stained with IL-12, TNFα, IFNγ or isotype antibodies. At least 1 × 105 gated events per condition were acquired. Data were acquired with BD FACSCanto II and analyzed using Tree Star FlowJo software.
DSS-Induced Murine Colitis Groups of C57BL/6 mice (n = 5/group) were inoculated orally with NCK2031, NCK56, NCK2025, or (5 × 108 CFU/100 μL PBS/mouse) for four consecutive days. In addition, a group of mice was treated with purified S. aureus- LTA (12.5 μg/μL) [27] for 5 days. All of these mice received one 5-day cycle of 3% DSS in drinking water, followed by 3 days of regular drinking water, with or without S. aureus- LTA, and then sacrificed on day 13. Acute colitis was observed after the first cycle of DSS in the non-inoculated group. Disease progression, including weight loss, diarrhea and fecal hemoccult blood positivity (FOB), was monitored throughout the study. Thereafter, mice were sacrificed and excised colon cross-sectional Swiss rolls were fixed in 10% formaldehyde and embedded in paraffin. Tissue sections (4 μm) were stained with hematoyxylin and eosin (H&E), and blindly scored, as described previously [28, 29]. The grading, based on a scale from 0 to 28, takes into account the degree of inflammatory infiltrate, the presence of erosion, ulceration, or necrosis, and the depth and surface extension of the lesion.

Results

Immune stimulation by NCK2031

To further characterize the role of LTA expression in inflammation, we attempted to generate s urface l ayer p rotein (Slp)-deficient strains using the upp-counterselective gene replacement system [20]. The resulting strain, NCK2031, showed no detectable expression of SlpB or SlpX while retaining LTA expression. Attempted insertional mutation in the slpA gene was unstable and spontaneous SlpA+ revertants occurred in the population. Therefore, the phenotype of this mutant was defined as SlpA+B-X-LTA+. SDS-PAGE analyses confirmed the expression of SlpA only (at 46 kDa), and the absence of SlpB and SlpX within the NCK2031 culture (data not shown). To evaluate any in vivo physiological effects of NCK2031 and its colonization in the gut of bacteria treated mice, the persistence of erythromycin-resistant (Emr) NCK56 and Emr NCK2031 strains in the colon were determined in C57BL/6 mice orally treated once with 5 × 108 CFU/mouse. Fecal pellets were collected the day before treatment and every day for up to 8 days after bacterial ingestion. Data reveal that mice cleared both NCK2031 and NCK56 (wild type) after 3 days, with significantly less NCK2031 excreted on days 2 and 3 (Figure 1a). Therefore, while the trends for clearance of the orally fed bacteria were similar, deletion of the SlpB and SlpX proteins appeared to accentuate clearance of the bacterium from the GI tract by one day, in vivo. Determining the time of bacterial clearance from the gut was essential to delineate the sampling time points used in the immunological studies.
To elucidate the physiological role of NCK2031 in dendritic cell (DC) activation, we co-cultured human monocyte derived DCs with NCK2031, NCK56, NCK2025, or purified S. aureus-LTA. Data revealed that the parent strain NCK56, the SlpA+B-X-LTA+ strain NCK2031, and S. aureus- LTA profoundly stimulated human DCs, while the SlpA+B+X+LTA- strain NCK2025 only marginally modified DC function (Figure 1b, c). Additionally, murine bone marrow derived DCs co-cultured with NCK2031 exhibited an up-regulation of MHC II, CD80, CD86, CD40, and increased production of intracellular interleukin (IL)-12 and TNFα when compared to DCs cultured with NCK56, NCK2025 or S. aureus- LTA (Figure 1d). These data indicate that LTA is not playing a regulatory role as seen in other studies [27, 30], but rather, functions as a highly pro-inflammatory molecule when expressed on L. acidophilus[19].

Activation of DCs, macrophages, and T cells by NCK2031

To overcome the undesired effects of cell death induced by enzymatic reagents (i.e. collagenase) necessary for cell isolation we employed TissueGnostics fluorescent microscopy [18] and confocal microscopy (Figure 2a, 3a) to analyze the cell populations and their cytokine expression in the colons of bacteria treated mice. As demonstrated previously, a significantly higher percentage of CD11c+IL-10+ DCs per total DAPI+ cells were observed in the colons of mice treated with NCK2025 when compared to mice treated with NCK2031, NCK56, or S. aureus- LTA on days 1 and 3, but not by day 7 (data not shown). In contrast, IL-12 and TNFα production by colonic DCs was significantly higher in mice treated with NCK2031, NCK56, or S. aureus-LTA when compared to mice that were treated with NCK2025 or PBS (Figure 2b). To expand upon these findings, we studied the ability of the LTA-expressing strains to induce the stimulation of DCs, macrophages, CD4+ and CD8+ T cells in vivo. While no significant differences were seen in the expression of MHC II, CD40, CD80, or CD86 on CD11c+CD11b+DCs or CD11b+F4/80+ macrophages (data not shown), cytoplasmic IL-12 and TNFα were significantly increased in CD11c+ MHC II+ DCs derived from the colons (Figure 2c), and mesenteric lymph nodes (MLNs) (Figure 3a), but not the spleens of mice that were treated with NCK2031, NCK56, or S. aureus -LTA versus PBS or NCK2025 (Figure 3b).
Recent studies have indicated that there is a significant recruitment of colonic F4/80+ macrophages in IBD compared to that seen in normal mucosa [31]. Thus, we investigated the impact of the genetically modified strains versus the wild type NCK56 and S. aureus-LTA on the recruitment and activation of colonic F4/80+ macrophages in vivo. Based on data generated by TissueGnostics, it was apparent that NCK2031, NCK56, and S. aureus-LTA significantly induced colonic F4/80+ macrophages to produce IL-12 and TNFα; the expression of these cytokines was low in NCK2025-treated mice after 24 hrs (Figure 4a, b), but had leveled off by days 3 and 7 (data not shown). In contrast, IL-10 was highly expressed in NCK2031, NCK56, NCK2025, and S. aureus- LTA treated mice compared to untreated mice (Figure 4c). This observation may reflect the nature of the bacteria and their gene products, which can differentially stimulate these colonic cells to become either pro-inflammatory or regulatory. These data suggest that activation of intestinal cells and induction of inflammation may be triggered by the presence of LTA, as previously shown in lungs with airway inflammation [32, 33].
In addition, colonic CD4+T cells from mice treated with NCK2031, NCK56, or S. aureus- LTA expressed higher levels of IFNγ compared to mice treated with NCK2025 or PBS (Figure 5a). Treatment of mice with NCK2031, NCK56, and S. aureus-LTA also induced significantly increased expression of IFNγ and TNFα (Figure 5b), but not IL-17 (data not shown), in the CD4+ T cells from the MLNs; a significant trend was not seen in the spleen (data not shown). The CD4+ cells seemed to be less affected in the NCK2025-treated mice (Figure 5b).
It has recently been demonstrated that CD8+T cells efficiently prevent the generation of colitis [34, 35] and play a critical role in controlling potentially pathogenic T cells [36] in IBD patients [37, 38]. Such CD8+T cells with regulatory activity reside in the lamina propria (LP) of healthy individuals but not in the LP of patients with IBD [37, 38]. Interestingly, the percentages of CD8+T cells in the colons of mice treated with NCK2031, NCK56, or S. aureus- LTA were significantly reduced (data not shown) when compared to NCK2025 treated mice. The majority of CD8+T cells in the colonic tissues from all groups of mice were IFNγ negative (data not shown), suggesting that colonic CD8+T cells are probably not involved in immune stimulation; rather, they exert immune regulation in the colon [36]. To demonstrate whether resident or recruited colonic CD8+ T cells are Foxp3+, these cells were visualized by TissueGnostics fluorescent microscopy. Surprisingly, there were no significant differences in Foxp3+ expression between the groups of the mice that were treated with the various L. acidophilus strains or S. aureus-LTA (data not shown). Further studies are required to study this cell type in colitis disease models. The activity of CD8+ T cells within the MLN and spleen was also analyzed. NCK2031, NCK56, and S. aureus-LTA significantly stimulated MLN CD8+CD44high T cells to produce IFNγ on day 3 and TNFα on day 7 (Figure 5c); no significant differences in the production of these cytokines were seen in the splenic CD8+ T cells (data not shown). Collectively, these data indicate that the LTA-expressing strains, NCK2031 and NCK56, activate DCs and macrophages in a pro-inflammatory manner, which in turn, may directly elicit CD4+ and CD8+ T cell activation.

Exacerbation of DSS-induced murine colitis by NCK2031 and S. aureus-LTA

To more thoroughly investigate the pro-inflammatory mechanisms induced by LTA, we utilized the DSS-mouse model for colitis [18]. Data show that DSS generated murine colitis in PBS-treated mice (Figure 6a-d). Mice began to lose weight after day 8 and developed severe diarrhea at days 10-13 (Figure 6b, c). To specifically address the role of LTA expression by NCK2031, mice were treated with NCK2031, NCK56, NCK2025 or S. aureus- LTA dissolved in drinking water. NCK2031, NCK56, or soluble S. aureus- LTA did not prevent the onset of colitis and the mice developed colitis similarly to non-treated mice (Figure 6a-d). In contrast, treatment with NCK2025 significantly prevented weight loss and reduced diarrhea (Figure 6), as previously documented [18]. Overall, the "Disease Activity Index" (DAI) was significantly increased for untreated animals and mice treated with NCK2031, NCK56, and S. aureus- LTA (Figure 6d). The colons from these mice exhibited ulcerated epithelium with significant inflammation confined to the mucosa, while colonic tissue obtained from mice treated with NCK2025 did not exhibit evidence of significant inflammation (Figure 6a).

Discussion

Excessive stimulation of intestinal innate cells, including DCs, with commensal bacteria and/or their gene products results in immune dysfunction that can generate uncontrolled inflammation leading to tissue destruction and colitis [39]. Precise cellular and molecular mechanisms of these induced inflammatory immune responses in IBD remain poorly understood. Data so far indicate that chronic intestinal inflammation coincides with elevated levels of pro-inflammatory cytokines (i.e., IL-12, TNFα) [40], and the differentiation/activation of pro-inflammatory DC-subsets and pathogenic CD4+T cells [41]. Accordingly, promising studies show that inhibition of detrimental signals induced by stimulatory bacterial products mitigates IBD progression [4249].
Together, our data show that LTA plays a critical role in the induction of the inflammatory responses and cannot prevent mice from developing colitis. Contrary to our findings, other studies have shown that lactobacilli-LTA induces regulatory signals (i.e., IL-10) via Erk1/2 signaling, resulting in anti-inflammatory mechanisms [30]. Other reports also indicate that commensal LTA protects mice from DSS-induced colitis and ameliorates impaired epithelial tight junctions [27, 50]. This discrepancy may lay in the methodological approaches and reagents that were used in these studies. Here we demonstrate that LTA and LTA expressing bacteria posses the ability to stimulate innate immune component cells, including DCs and macrophages, which in turn, can trigger pathogenic IFNγ-secreting CD4+ T cells in disease progression.
The role of IFNγ expressed in the colon is unclear, as it has been shown that this cytokine exerts protective features in different models of inflammation [51]. For example, in a murine model of multiple sclerosis, neutralization of IFNγ resulted in inflammatory immune responses [52]. Similarly, in a mouse model of IBD, IFNγ has been shown to suppress IL-23 [53], and collagen-induced arthritis is induced in the absence of IFNγ signaling [54]. However, in the models we have previously used for experimental DSS inducing epithelial injury (i.e., leaky mucosa) resulting in colitis or pathogenic CD4+CD45RBhigh T cell transfer; IFNγ seems to be involved in the pathogenesis of the induced colitis. Additionally, IFNγ contributes to intestinal pathology and lethality in lipopolysaccharide-sensitization models of toxic shock syndrome (TSS) [55, 56]. Further studies are needed to clearly elucidate the role of this cytokine in colitis and the timing of its production during disease progression.
Our results show that in vitro and in vivo, NCK2031 expressing LTA and purified S. aureus-LTA induce intestinal immune activation resulting in the production of TNFα and IL-12 and the development of pro-inflammatory cells such as CD11c+TNFα+IL-12+ DCs and F4/80+ macrophages that may contribute to an undesired CD4+IFNγ+TNFα+ T cell pro-inflammatory response seen in colitis. Conversely, treatment with LTA-deficient NCK2025 elicited regulatory signals and induced less inflammation, as demonstrated here and previously [18, 19]. We have previously demonstrated the involvement of SlpA of L. acidophilus NCFM in the immune regulation of DC functions [57]. Studies are underway to better delineate the physiological role of SlpA from this bacterium in vivo. Finally, an acute colitis characterized by bloody diarrhea, ulcerations and infiltrations with granulocytes can be induced in mice that are exposed for several days to significantly dissolved DSS polymers in drinking water [58, 59]. As seen in Figure 6, we also could confirm such clinical signs when DSS was used to induce colitis. Additionally, our data clearly show not only the proinflammatory nature of LTA but also the significantly exacerbated disease progression in the colon of mice that received LTA in the drinking water. Thus, these data are in sharp contrast to the notion that LTA plays a regulatory role and possesses the ability to mitigate DSS-induced colitis, as described previously [27]. In summary, oral administration of DSS to mice induces an acute colitis, followed by a slow colonic regeneration of the epithelium with a concomitant exaggerated inflammation; furthermore, LTA did not reduce the levels DSS-induced colitis or promote the rapid healing of ulcerated and destroyed epithelial tissues in our studies.

Conclusions

Our data strongly suggest that directed alteration of two of the L. acidophilus-Slps did not ameliorate LTA-induced pro-inflammatory signals and subsequent colitis. Additionally, further studies are required to elucidate the role of L. acidophilus-Slps in the steady state of normal gut homeostasis and in diseases such as colitis and colon cancer where deregulated inflammation plays a critical role in disease progression. Finally, LTA did not mitigate DSS-induced colitis but induced further colonic inflammation.

Acknowledgements

This work was supported in part by the by NIH Grant 1R01AI098833-01, Danisco USA, and the North Carolina Dairy Foundation. We also thank Dr. Praveen Bere, Dr. Mary Brown, and Robert Roman for their generous support and excellent technical assistant and discussion of this work.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MZ and MWK carried out all animal work, immunocytochemistry (i.e., high throughput and confocal imaging) and flow cytometry experiments. Additionally, MZ and MWK gathered the raw data and using statistical analyses and graphic programs compiled the data into meaningful information. MZ, MWK, JLO also contributed to writing the Methods section of the manuscript. MZ and MWK collected all of the graphics and formed the figures of the Manuscript. YJG, TK designed experiments and contributed to writing the Methods section. KS performed Slp proteome analysis on NCK2031 strain. MM designed experiments, discussed and wrote the manuscript. All authors have read and approved this manuscript.
Literatur
2.
Zurück zum Zitat Lee YK, Mazmanian SK: Has the microbiota played a critical role in the evolution of the adaptive immune system?. Science. 2010, 330: 1768-1773. 10.1126/science.1195568.PubMedCentralCrossRefPubMed Lee YK, Mazmanian SK: Has the microbiota played a critical role in the evolution of the adaptive immune system?. Science. 2010, 330: 1768-1773. 10.1126/science.1195568.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Powrie F: T cells in inflammatory bowel disease: protective and pathogenic roles. Immunity. 1995, 3: 171-174. 10.1016/1074-7613(95)90086-1.CrossRefPubMed Powrie F: T cells in inflammatory bowel disease: protective and pathogenic roles. Immunity. 1995, 3: 171-174. 10.1016/1074-7613(95)90086-1.CrossRefPubMed
5.
Zurück zum Zitat Cheroutre H: In IBD eight can come before four. Gastroenterology. 2006, 131: 667-670. 10.1053/j.gastro.2006.06.041.CrossRefPubMed Cheroutre H: In IBD eight can come before four. Gastroenterology. 2006, 131: 667-670. 10.1053/j.gastro.2006.06.041.CrossRefPubMed
6.
Zurück zum Zitat Elson CO, Cong Y, McCracken VJ, Dimmitt RA, Lorenz RG, Weaver CT: Experimental models of inflammatory bowel disease reveal innate, adaptive, and regulatory mechanisms of host dialogue with the microbiota. Immunol Rev. 2005, 206: 260-276. 10.1111/j.0105-2896.2005.00291.x.CrossRefPubMed Elson CO, Cong Y, McCracken VJ, Dimmitt RA, Lorenz RG, Weaver CT: Experimental models of inflammatory bowel disease reveal innate, adaptive, and regulatory mechanisms of host dialogue with the microbiota. Immunol Rev. 2005, 206: 260-276. 10.1111/j.0105-2896.2005.00291.x.CrossRefPubMed
7.
Zurück zum Zitat Coombes JL, Robinson NJ, Maloy KJ, Uhlig HH, Powrie F: Regulatory T cells and intestinal homeostasis. Immunol Rev. 2005, 204: 184-194. 10.1111/j.0105-2896.2005.00250.x.CrossRefPubMed Coombes JL, Robinson NJ, Maloy KJ, Uhlig HH, Powrie F: Regulatory T cells and intestinal homeostasis. Immunol Rev. 2005, 204: 184-194. 10.1111/j.0105-2896.2005.00250.x.CrossRefPubMed
8.
Zurück zum Zitat Read S, Malmstrom V, Powrie F: Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000, 192: 295-302. 10.1084/jem.192.2.295.PubMedCentralCrossRefPubMed Read S, Malmstrom V, Powrie F: Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000, 192: 295-302. 10.1084/jem.192.2.295.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Fontenot JD, Gavin MA, Rudensky AY: Foxp3 programs the development and function of CD4 + CD25+ regulatory T cells. Nat Immunol. 2003, 4: 330-336.CrossRefPubMed Fontenot JD, Gavin MA, Rudensky AY: Foxp3 programs the development and function of CD4 + CD25+ regulatory T cells. Nat Immunol. 2003, 4: 330-336.CrossRefPubMed
10.
11.
Zurück zum Zitat Alkan ML, Beachey EH: Excretion of lipoteichoic acid by group A streptococci. Influence of penicillin on excretion and loss of ability to adhere to human oral mucosal cells. J Clin Invest. 1978, 61: 671-677. 10.1172/JCI108979.PubMedCentralCrossRefPubMed Alkan ML, Beachey EH: Excretion of lipoteichoic acid by group A streptococci. Influence of penicillin on excretion and loss of ability to adhere to human oral mucosal cells. J Clin Invest. 1978, 61: 671-677. 10.1172/JCI108979.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Reichmann NT, Grundling A: Location, synthesis and function of glycolipids and polyglycerolphosphate lipoteichoic acid in Gram-positive bacteria of the phylum Firmicutes. FEMS MicrobiolLett. 2011, 319: 97-105. 10.1111/j.1574-6968.2011.02260.x.CrossRef Reichmann NT, Grundling A: Location, synthesis and function of glycolipids and polyglycerolphosphate lipoteichoic acid in Gram-positive bacteria of the phylum Firmicutes. FEMS MicrobiolLett. 2011, 319: 97-105. 10.1111/j.1574-6968.2011.02260.x.CrossRef
13.
Zurück zum Zitat Su SC, Hua KF, Lee H, Chao LK, Tan SK, Yang SF, et al: LTA and LPS mediated activation of protein kinases in the regulation of inflammatory cytokines expression in macrophages. ClinChimActa. 2006, 374: 106-115. Su SC, Hua KF, Lee H, Chao LK, Tan SK, Yang SF, et al: LTA and LPS mediated activation of protein kinases in the regulation of inflammatory cytokines expression in macrophages. ClinChimActa. 2006, 374: 106-115.
14.
Zurück zum Zitat Zidek Z, Farghali H, Kmonickova E: Intrinsic nitric oxide-stimulatory activity of lipoteichoic acids from different Gram-positive bacteria. Nitric Oxide. 2010, 23: 300-310. 10.1016/j.niox.2010.09.001.CrossRefPubMed Zidek Z, Farghali H, Kmonickova E: Intrinsic nitric oxide-stimulatory activity of lipoteichoic acids from different Gram-positive bacteria. Nitric Oxide. 2010, 23: 300-310. 10.1016/j.niox.2010.09.001.CrossRefPubMed
15.
Zurück zum Zitat Chang HC, Lin KH, Tai YT, Chen JT, Chen RM: Lipoteichoic acid-induced TNF-alpha and IL-6 gene expressions and oxidative stress production in macrophages are suppressed by ketamine through downregulating Toll-like receptor 2-mediated activation oF ERK1/2 and NFkappaB. Shock. 2010, 33: 485-492.CrossRefPubMed Chang HC, Lin KH, Tai YT, Chen JT, Chen RM: Lipoteichoic acid-induced TNF-alpha and IL-6 gene expressions and oxidative stress production in macrophages are suppressed by ketamine through downregulating Toll-like receptor 2-mediated activation oF ERK1/2 and NFkappaB. Shock. 2010, 33: 485-492.CrossRefPubMed
16.
Zurück zum Zitat Kengatharan KM, De Kimpe S, Robson C, Foster SJ, Thiemermann C: Mechanism of gram-positive shock: identification of peptidoglycan and lipoteichoic acid moieties essential in the induction of nitric oxide synthase, shock, and multiple organ failure. J Exp Med. 1998, 188: 305-315. 10.1084/jem.188.2.305.PubMedCentralCrossRefPubMed Kengatharan KM, De Kimpe S, Robson C, Foster SJ, Thiemermann C: Mechanism of gram-positive shock: identification of peptidoglycan and lipoteichoic acid moieties essential in the induction of nitric oxide synthase, shock, and multiple organ failure. J Exp Med. 1998, 188: 305-315. 10.1084/jem.188.2.305.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Akira S, Uematsu S, Takeuchi O: Pathogen recognition and innate immunity. Cell. 2006, 124: 783-801. 10.1016/j.cell.2006.02.015.CrossRefPubMed Akira S, Uematsu S, Takeuchi O: Pathogen recognition and innate immunity. Cell. 2006, 124: 783-801. 10.1016/j.cell.2006.02.015.CrossRefPubMed
18.
Zurück zum Zitat Mohamadzadeh M, Pfeiler EA, Brown JB, Zadeh M, Gramarossa M, Managlia E, et al: Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc NatlAcadSci USA. 2011, 108: 4623-4630. 10.1073/pnas.1005066107.CrossRef Mohamadzadeh M, Pfeiler EA, Brown JB, Zadeh M, Gramarossa M, Managlia E, et al: Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc NatlAcadSci USA. 2011, 108: 4623-4630. 10.1073/pnas.1005066107.CrossRef
19.
Zurück zum Zitat Saber R, Zadeh M, Pakanati KC, Bere P, Klaenhammer T, Mohamadzadeh M: Lipoteichoic acid-deficient Lactobacillus acidophilus regulates downstream signals. Immunotherapy. 2011, 3: 337-347. 10.2217/imt.10.119.CrossRefPubMed Saber R, Zadeh M, Pakanati KC, Bere P, Klaenhammer T, Mohamadzadeh M: Lipoteichoic acid-deficient Lactobacillus acidophilus regulates downstream signals. Immunotherapy. 2011, 3: 337-347. 10.2217/imt.10.119.CrossRefPubMed
20.
Zurück zum Zitat Goh YJ, Azcarate-Peril MA, O'Flaherty S, Durmaz E, Valence F, Jardin J, et al: Development and application of a upp-based counterselective gene replacement system for the study of the S-layer protein SlpX of Lactobacillus acidophilus NCFM. Appl Environ Microbiol. 2009, 75: 3093-3105. 10.1128/AEM.02502-08.PubMedCentralCrossRefPubMed Goh YJ, Azcarate-Peril MA, O'Flaherty S, Durmaz E, Valence F, Jardin J, et al: Development and application of a upp-based counterselective gene replacement system for the study of the S-layer protein SlpX of Lactobacillus acidophilus NCFM. Appl Environ Microbiol. 2009, 75: 3093-3105. 10.1128/AEM.02502-08.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Russell WM, Klaenhammer TR: Efficient system for directed integration into the Lactobacillus acidophilus and Lactobacillus gasseri chromosomes via homologous recombination. Appl Environ Microbiol. 2001, 67: 4361-4364. 10.1128/AEM.67.9.4361-4364.2001.PubMedCentralCrossRefPubMed Russell WM, Klaenhammer TR: Efficient system for directed integration into the Lactobacillus acidophilus and Lactobacillus gasseri chromosomes via homologous recombination. Appl Environ Microbiol. 2001, 67: 4361-4364. 10.1128/AEM.67.9.4361-4364.2001.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Mohamadzadeh M, Berard F, Essert G, Chalouni C, Pulendran B, Davoust J, et al: Interleukin 15 skews monocyte differentiation into dendritic cells with features of Langerhans cells. J Exp Med. 2001, 194: 1013-1020. 10.1084/jem.194.7.1013.PubMedCentralCrossRefPubMed Mohamadzadeh M, Berard F, Essert G, Chalouni C, Pulendran B, Davoust J, et al: Interleukin 15 skews monocyte differentiation into dendritic cells with features of Langerhans cells. J Exp Med. 2001, 194: 1013-1020. 10.1084/jem.194.7.1013.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Sallusto F, Lanzavecchia A: Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994, 179: 1109-1118. 10.1084/jem.179.4.1109.CrossRefPubMed Sallusto F, Lanzavecchia A: Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994, 179: 1109-1118. 10.1084/jem.179.4.1109.CrossRefPubMed
24.
Zurück zum Zitat Pulendran B, Dillon S, Joseph C, Curiel T, Banchereau J, Mohamadzadeh M: Dendritic cells generated in the presence of GM-CSF plus IL-15 prime potent CD8+ Tc1 responses in vivo. Eur J Immunol. 2004, 34: 66-73. 10.1002/eji.200324567.CrossRefPubMed Pulendran B, Dillon S, Joseph C, Curiel T, Banchereau J, Mohamadzadeh M: Dendritic cells generated in the presence of GM-CSF plus IL-15 prime potent CD8+ Tc1 responses in vivo. Eur J Immunol. 2004, 34: 66-73. 10.1002/eji.200324567.CrossRefPubMed
25.
Zurück zum Zitat Gounaris E, Tung CH, Restaino C, Maehr R, Kohler R, Joyce JA, et al: Live imaging of cysteine-cathepsin activity reveals dynamics of focal inflammation, angiogenesis, and polyp growth. PLoS One. 2008, 3: e2916-10.1371/journal.pone.0002916.PubMedCentralCrossRefPubMed Gounaris E, Tung CH, Restaino C, Maehr R, Kohler R, Joyce JA, et al: Live imaging of cysteine-cathepsin activity reveals dynamics of focal inflammation, angiogenesis, and polyp growth. PLoS One. 2008, 3: e2916-10.1371/journal.pone.0002916.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Mohamadzadeh M, Olson S, Kalina WV, Ruthel G, Demmin GL, Warfield KL, et al: Lactobacilli activate human dendritic cells that skew T cells toward T helper 1 polarization. Proc NatlAcadSci USA. 2005, 02 (8): 2880-2885.CrossRef Mohamadzadeh M, Olson S, Kalina WV, Ruthel G, Demmin GL, Warfield KL, et al: Lactobacilli activate human dendritic cells that skew T cells toward T helper 1 polarization. Proc NatlAcadSci USA. 2005, 02 (8): 2880-2885.CrossRef
27.
Zurück zum Zitat Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R: Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004, 118: 229-241. 10.1016/j.cell.2004.07.002.CrossRefPubMed Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R: Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004, 118: 229-241. 10.1016/j.cell.2004.07.002.CrossRefPubMed
28.
Zurück zum Zitat Cooper HS, Murthy SN, Shah RS, Sedergran DJ: Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 1993, 69: 238-249.PubMed Cooper HS, Murthy SN, Shah RS, Sedergran DJ: Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 1993, 69: 238-249.PubMed
29.
Zurück zum Zitat Murthy SN, Cooper HS, Shim H, Shah RS, Ibrahim SA, Sedergran DJ: Treatment of dextran sulfate sodium-induced murine colitis by intracoloniccyclosporin. Dig Dis Sci. 1993, 38: 1722-1734. 10.1007/BF01303184.CrossRefPubMed Murthy SN, Cooper HS, Shim H, Shah RS, Ibrahim SA, Sedergran DJ: Treatment of dextran sulfate sodium-induced murine colitis by intracoloniccyclosporin. Dig Dis Sci. 1993, 38: 1722-1734. 10.1007/BF01303184.CrossRefPubMed
30.
Zurück zum Zitat Kaji R, Kiyoshima-Shibata J, Nagaoka M, Nanno M, Shida K: Bacterial teichoic acids reverse predominant IL-12 production induced by certain lactobacillus strains into predominant IL-10 production via TLR2-dependent ERK activation in macrophages. J Immunol. 2010, 184: 3505-3513. 10.4049/jimmunol.0901569.CrossRefPubMed Kaji R, Kiyoshima-Shibata J, Nagaoka M, Nanno M, Shida K: Bacterial teichoic acids reverse predominant IL-12 production induced by certain lactobacillus strains into predominant IL-10 production via TLR2-dependent ERK activation in macrophages. J Immunol. 2010, 184: 3505-3513. 10.4049/jimmunol.0901569.CrossRefPubMed
31.
Zurück zum Zitat Lloyd CM, Phillips AR, Cooper GJ, Dunbar PR: Three-colour fluorescence immunohistochemistry reveals the diversity of cells staining for macrophage markers in murine spleen and liver. J Immunol Methods. 2008, 334: 70-81. 10.1016/j.jim.2008.02.005.CrossRefPubMed Lloyd CM, Phillips AR, Cooper GJ, Dunbar PR: Three-colour fluorescence immunohistochemistry reveals the diversity of cells staining for macrophage markers in murine spleen and liver. J Immunol Methods. 2008, 334: 70-81. 10.1016/j.jim.2008.02.005.CrossRefPubMed
32.
Zurück zum Zitat Lee IT, Lee CW, Tung WH, Wang SW, Lin CC, Shu JC, et al: Cooperation of TLR2 with MyD88, PI3K, and Rac1 in lipoteichoic acid-induced cPLA2/COX-2-dependent airway inflammatory responses. Am J Pathol. 2010, 176: 1671-1684. 10.2353/ajpath.2010.090714.PubMedCentralCrossRefPubMed Lee IT, Lee CW, Tung WH, Wang SW, Lin CC, Shu JC, et al: Cooperation of TLR2 with MyD88, PI3K, and Rac1 in lipoteichoic acid-induced cPLA2/COX-2-dependent airway inflammatory responses. Am J Pathol. 2010, 176: 1671-1684. 10.2353/ajpath.2010.090714.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Lee IT, Wang SW, Lee CW, Chang CC, Lin CC, Luo SF, et al: Lipoteichoic acid induces HO-1 expression via the TLR2/MyD88/c-Src/NADPH oxidase pathway and Nrf2 in human tracheal smooth muscle cells. J Immunol. 2008, 181: 5098-5110.CrossRefPubMed Lee IT, Wang SW, Lee CW, Chang CC, Lin CC, Luo SF, et al: Lipoteichoic acid induces HO-1 expression via the TLR2/MyD88/c-Src/NADPH oxidase pathway and Nrf2 in human tracheal smooth muscle cells. J Immunol. 2008, 181: 5098-5110.CrossRefPubMed
34.
Zurück zum Zitat Menager-Marcq I, Pomie C, Romagnoli P, van Meerwijk JP: CD8 + CD28- regulatory T lymphocytes prevent experimental inflammatory bowel disease in mice. Gastroenterology. 2006, 131: 1775-1785. 10.1053/j.gastro.2006.09.008.PubMedCentralCrossRefPubMed Menager-Marcq I, Pomie C, Romagnoli P, van Meerwijk JP: CD8 + CD28- regulatory T lymphocytes prevent experimental inflammatory bowel disease in mice. Gastroenterology. 2006, 131: 1775-1785. 10.1053/j.gastro.2006.09.008.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Ho J, Kurtz CC, Naganuma M, Ernst PB, Cominelli F, Rivera-Nieves J: A CD8+/CD103high T cell subset regulates TNF-mediated chronic murine ileitis. J Immunol. 2008, 180: 2573-2580.PubMedCentralCrossRefPubMed Ho J, Kurtz CC, Naganuma M, Ernst PB, Cominelli F, Rivera-Nieves J: A CD8+/CD103high T cell subset regulates TNF-mediated chronic murine ileitis. J Immunol. 2008, 180: 2573-2580.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Fleissner D, Frede A, Knott M, Knuschke T, Geffers R, Hansen W, et al: Generation and function of immunosuppressive human and murine CD8(+) T cells by transforming growth factor-beta and retinoic acid. Immunol. 2011, 134: 82-92. 10.1111/j.1365-2567.2011.03469.x.CrossRef Fleissner D, Frede A, Knott M, Knuschke T, Geffers R, Hansen W, et al: Generation and function of immunosuppressive human and murine CD8(+) T cells by transforming growth factor-beta and retinoic acid. Immunol. 2011, 134: 82-92. 10.1111/j.1365-2567.2011.03469.x.CrossRef
37.
Zurück zum Zitat Allez M, Brimnes J, Dotan I, Mayer L: Expansion of CD8+ T cells with regulatory function after interaction with intestinal epithelial cells. Gastroenterology. 2002, 123: 1516-1526. 10.1053/gast.2002.36588.CrossRefPubMed Allez M, Brimnes J, Dotan I, Mayer L: Expansion of CD8+ T cells with regulatory function after interaction with intestinal epithelial cells. Gastroenterology. 2002, 123: 1516-1526. 10.1053/gast.2002.36588.CrossRefPubMed
38.
Zurück zum Zitat Brimnes J, Allez M, Dotan I, Shao L, Nakazawa A, Mayer L: Defects in CD8+ regulatory T cells in the lamina propria of patients with inflammatory bowel disease. J Immunol. 2005, 174: 5814-5822.CrossRefPubMed Brimnes J, Allez M, Dotan I, Shao L, Nakazawa A, Mayer L: Defects in CD8+ regulatory T cells in the lamina propria of patients with inflammatory bowel disease. J Immunol. 2005, 174: 5814-5822.CrossRefPubMed
39.
Zurück zum Zitat Macdonald TT, Monteleone G: Immunity, inflammation, and allergy in the gut. Science. 2005, 307: 1920-1925. 10.1126/science.1106442.CrossRefPubMed Macdonald TT, Monteleone G: Immunity, inflammation, and allergy in the gut. Science. 2005, 307: 1920-1925. 10.1126/science.1106442.CrossRefPubMed
40.
Zurück zum Zitat Rutgeerts P, Vermeire S, Van Assche G: Biological therapies for inflammatory bowel diseases. Gastroenterology. 2009, 136: 1182-1197. 10.1053/j.gastro.2009.02.001.CrossRefPubMed Rutgeerts P, Vermeire S, Van Assche G: Biological therapies for inflammatory bowel diseases. Gastroenterology. 2009, 136: 1182-1197. 10.1053/j.gastro.2009.02.001.CrossRefPubMed
41.
Zurück zum Zitat Strober W, Fuss IJ, Blumberg RS: The immunology of mucosal models of inflammation. Annu Rev Immunol. 2002, 20: 495-549. 10.1146/annurev.immunol.20.100301.064816.CrossRefPubMed Strober W, Fuss IJ, Blumberg RS: The immunology of mucosal models of inflammation. Annu Rev Immunol. 2002, 20: 495-549. 10.1146/annurev.immunol.20.100301.064816.CrossRefPubMed
42.
Zurück zum Zitat Simpson SJ, Shah S, Comiskey M, de Jong YP, Wang B, Mizoguchi E, et al: T cell-mediated pathology in two models of experimental colitis depends predominantly on the interleukin 12/Signal transducer and activator of transcription (Stat)-4 pathway, but is not conditional on interferon gamma expression by T cells. J Exp Med. 1998, 187: 1225-1234. 10.1084/jem.187.8.1225.PubMedCentralCrossRefPubMed Simpson SJ, Shah S, Comiskey M, de Jong YP, Wang B, Mizoguchi E, et al: T cell-mediated pathology in two models of experimental colitis depends predominantly on the interleukin 12/Signal transducer and activator of transcription (Stat)-4 pathway, but is not conditional on interferon gamma expression by T cells. J Exp Med. 1998, 187: 1225-1234. 10.1084/jem.187.8.1225.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Kullberg MC, Rothfuchs AG, Jankovic D, Caspar P, Wynn TA, Gorelick PL, et al: Helicobacter hepaticus-induced colitis in interleukin-10-deficient mice: cytokine requirements for the induction and maintenance of intestinal inflammation. Infect Immun. 2001, 69: 4232-4241. 10.1128/IAI.69.7.4232-4241.2001.PubMedCentralCrossRefPubMed Kullberg MC, Rothfuchs AG, Jankovic D, Caspar P, Wynn TA, Gorelick PL, et al: Helicobacter hepaticus-induced colitis in interleukin-10-deficient mice: cytokine requirements for the induction and maintenance of intestinal inflammation. Infect Immun. 2001, 69: 4232-4241. 10.1128/IAI.69.7.4232-4241.2001.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Monteleone I, Sarra M, Del Vecchio Blanco G, Paoluzi OA, Franze E, Fina D, et al: Characterization of IL-17A-producing cells in celiac disease mucosa. J Immunol. 2010, 184: 2211-2218. 10.4049/jimmunol.0901919.CrossRefPubMed Monteleone I, Sarra M, Del Vecchio Blanco G, Paoluzi OA, Franze E, Fina D, et al: Characterization of IL-17A-producing cells in celiac disease mucosa. J Immunol. 2010, 184: 2211-2218. 10.4049/jimmunol.0901919.CrossRefPubMed
45.
Zurück zum Zitat Pallone F, Fina D, Caruso R, Monteleone G: Role of IL-21 in inflammatory bowel disease. Expert Rev ClinImmunol. 2010, 6: 537-541.CrossRef Pallone F, Fina D, Caruso R, Monteleone G: Role of IL-21 in inflammatory bowel disease. Expert Rev ClinImmunol. 2010, 6: 537-541.CrossRef
46.
47.
Zurück zum Zitat Sarra M, Monteleone I, Stolfi C, Fantini MC, Sileri P, Sica G, et al: Interferon-gamma-expressing cells are a major source of interleukin-21 in inflammatory bowel diseases. Inflamm Bowel Dis. 2010, 16: 1332-1339.CrossRefPubMed Sarra M, Monteleone I, Stolfi C, Fantini MC, Sileri P, Sica G, et al: Interferon-gamma-expressing cells are a major source of interleukin-21 in inflammatory bowel diseases. Inflamm Bowel Dis. 2010, 16: 1332-1339.CrossRefPubMed
48.
Zurück zum Zitat Sarra M, Pallone F, Macdonald TT, Monteleone G: IL-23/IL-17 axis in IBD. Inflamm Bowel Dis. 2010, 16: 1808-1813. 10.1002/ibd.21248.CrossRefPubMed Sarra M, Pallone F, Macdonald TT, Monteleone G: IL-23/IL-17 axis in IBD. Inflamm Bowel Dis. 2010, 16: 1808-1813. 10.1002/ibd.21248.CrossRefPubMed
49.
Zurück zum Zitat Popivanova BK, Kitamura K, Wu Y, Kondo T, Kagaya T, Kaneko S, et al: Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J Clin Invest. 2008, 118: 560-570.PubMedCentralPubMed Popivanova BK, Kitamura K, Wu Y, Kondo T, Kagaya T, Kaneko S, et al: Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J Clin Invest. 2008, 118: 560-570.PubMedCentralPubMed
50.
Zurück zum Zitat Miyauchi E, Morita H, Okuda J, Sashihara T, Shimizu M, Tanabe S: Cell wall fraction of Enterococcus hirae ameliorates TNF-alpha-induced barrier impairment in the human epithelial tight junction. LettApplMicrobiol. 2008, 46: 469-476. Miyauchi E, Morita H, Okuda J, Sashihara T, Shimizu M, Tanabe S: Cell wall fraction of Enterococcus hirae ameliorates TNF-alpha-induced barrier impairment in the human epithelial tight junction. LettApplMicrobiol. 2008, 46: 469-476.
51.
Zurück zum Zitat Korn T, Bettelli E, Oukka M, Kuchroo VK: IL-17 and Th17 Cells. Annu Rev Immunol. 2009, 27: 485-517. 10.1146/annurev.immunol.021908.132710.CrossRefPubMed Korn T, Bettelli E, Oukka M, Kuchroo VK: IL-17 and Th17 Cells. Annu Rev Immunol. 2009, 27: 485-517. 10.1146/annurev.immunol.021908.132710.CrossRefPubMed
52.
Zurück zum Zitat Billiau A, Heremans H, Vandekerckhove F, Dijkmans R, Sobis H, Meulepas E, et al: Enhancement of experimental allergic encephalomyelitis in mice by antibodies against IFN-gamma. J Immunol. 1988, 140: 1506-1510.PubMed Billiau A, Heremans H, Vandekerckhove F, Dijkmans R, Sobis H, Meulepas E, et al: Enhancement of experimental allergic encephalomyelitis in mice by antibodies against IFN-gamma. J Immunol. 1988, 140: 1506-1510.PubMed
53.
Zurück zum Zitat Sheikh SZ, Matsuoka K, Kobayashi T, Li F, Rubinas T, Plevy SE: Cutting edge: IFN-gamma is a negative regulator of IL-23 in murine macrophages and experimental colitis. J Immunol. 2010, 184: 4069-4073. 10.4049/jimmunol.0903600.PubMedCentralCrossRefPubMed Sheikh SZ, Matsuoka K, Kobayashi T, Li F, Rubinas T, Plevy SE: Cutting edge: IFN-gamma is a negative regulator of IL-23 in murine macrophages and experimental colitis. J Immunol. 2010, 184: 4069-4073. 10.4049/jimmunol.0903600.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Manoury-Schwartz B, Chiocchia G, Bessis N, Abehsira-Amar O, Batteux F, Muller S, et al: High susceptibility to collagen-induced arthritis in mice lacking IFN-gamma receptors. J Immunol. 1997, 158: 5501-5506.PubMed Manoury-Schwartz B, Chiocchia G, Bessis N, Abehsira-Amar O, Batteux F, Muller S, et al: High susceptibility to collagen-induced arthritis in mice lacking IFN-gamma receptors. J Immunol. 1997, 158: 5501-5506.PubMed
55.
Zurück zum Zitat Dinges MM, Schlievert PM: Role of T cells and gamma interferon during induction of hypersensitivity to lipopolysaccharide by toxic shock syndrome toxin 1 in mice. Infect Immun. 2001, 69: 1256-1264. 10.1128/IAI.69.3.1256-1264.2001.PubMedCentralCrossRefPubMed Dinges MM, Schlievert PM: Role of T cells and gamma interferon during induction of hypersensitivity to lipopolysaccharide by toxic shock syndrome toxin 1 in mice. Infect Immun. 2001, 69: 1256-1264. 10.1128/IAI.69.3.1256-1264.2001.PubMedCentralCrossRefPubMed
56.
Zurück zum Zitat Tilahun AY, Holz M, Wu TT, David CS, Rajagopalan G: Interferon gamma-dependent intestinal pathology contributes to the lethality in bacterial superantigen-induced toxic shock syndrome. PLoS One. 2011, 6: e16764-10.1371/journal.pone.0016764.PubMedCentralCrossRefPubMed Tilahun AY, Holz M, Wu TT, David CS, Rajagopalan G: Interferon gamma-dependent intestinal pathology contributes to the lethality in bacterial superantigen-induced toxic shock syndrome. PLoS One. 2011, 6: e16764-10.1371/journal.pone.0016764.PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Konstantinov SR, Smidt H, de Vos WM, Bruijns SC, Singh SK, Valence F, et al: S layer protein A of Lactobacillus acidophilus NCFM regulates immature dendritic cell and T cell functions. Proc NatlAcadSci USA. 2008, 105: 19474-19479. 10.1073/pnas.0810305105.CrossRef Konstantinov SR, Smidt H, de Vos WM, Bruijns SC, Singh SK, Valence F, et al: S layer protein A of Lactobacillus acidophilus NCFM regulates immature dendritic cell and T cell functions. Proc NatlAcadSci USA. 2008, 105: 19474-19479. 10.1073/pnas.0810305105.CrossRef
58.
Zurück zum Zitat Dieleman LA, Palmen MJ, Akol H, Bloemena E, Pena AS, Meuwissen SG, et al: Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin Exp Immunol. 1998, 114: 385-391. 10.1046/j.1365-2249.1998.00728.x.PubMedCentralCrossRefPubMed Dieleman LA, Palmen MJ, Akol H, Bloemena E, Pena AS, Meuwissen SG, et al: Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin Exp Immunol. 1998, 114: 385-391. 10.1046/j.1365-2249.1998.00728.x.PubMedCentralCrossRefPubMed
59.
Zurück zum Zitat Wirtz S, Neufert C, Weigmann B, Neurath MF: Chemically induced mouse models of intestinal inflammation. Nat Protoc. 2007, 2: 541-546.CrossRefPubMed Wirtz S, Neufert C, Weigmann B, Neurath MF: Chemically induced mouse models of intestinal inflammation. Nat Protoc. 2007, 2: 541-546.CrossRefPubMed
Metadaten
Titel
Induction of intestinal pro-inflammatory immune responses by lipoteichoic acid
verfasst von
Mojgan Zadeh
Mohammad W Khan
Yong Jun Goh
Kurt Selle
Jennifer L Owen
Todd Klaenhammer
Mansour Mohamadzadeh
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Journal of Inflammation / Ausgabe 1/2012
Elektronische ISSN: 1476-9255
DOI
https://doi.org/10.1186/1476-9255-9-7

Weitere Artikel der Ausgabe 1/2012

Journal of Inflammation 1/2012 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.